Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20.531
Filter
1.
Sci Rep ; 14(1): 15143, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956228

ABSTRACT

Laboratory mice are typically housed in "shoebox" cages with limited opportunities to engage in natural behaviour. Temporary access to environments with increased space and complexity (playpens) may improve mouse welfare. Previous work by our group has shown that mice are motivated to access and use these environments, but it is unknown how other aspects of welfare are impacted. Female C57BL/6J, BALB/cJ, and DBA/2J mice (n = 21; 7 mice per strain) were housed in mixed-strain trios and given temporary access to a large playpen with their cage mates three times per week. Control mice (n = 21; 7 mice per strain) remained in their home cages. Home cage behaviour (development of stereotypic behaviour over time, aggression following cage-changing) and anxiety tests were used to assess how playpen access impacted welfare. Contrary to our predictions, we found increased time spent performing stereotypies in playpen mice; this difference may be related to negative emotional states, increased motivation to escape the home cage, or active coping strategies. Playpen access resulted in strain-dependent improvements in aggression and some measures of anxiety. Aggression was lower for C57BL/6J mice in the playpen treatment following cage changing than it was for C57BL/6J control mice, while playpen mice, and particularly the C57BL/6J strain, spent more time in the center of the open field test and produced fewer fecal boli during anxiety testing, supporting other research showing that strain differences play an important role in behaviour and stress resiliency.


Subject(s)
Aggression , Animal Welfare , Behavior, Animal , Housing, Animal , Mice, Inbred C57BL , Animals , Mice , Female , Behavior, Animal/physiology , Anxiety , Mice, Inbred BALB C , Mice, Inbred DBA , Stereotyped Behavior
2.
Front Immunol ; 15: 1381802, 2024.
Article in English | MEDLINE | ID: mdl-38966637

ABSTRACT

Background: Yishen-Tongbi Decoction (YSTB), a traditional Chinese prescription, has been used to improve syndromes of rheumatoid arthritis (RA) for many years. Previous research has shown that YSTB has anti-inflammatory and analgesic properties. However, the underlying molecular mechanism of the anti-RA effects of YSTB remains unclear. Purpose and study design: The purpose of this research was to investigate how YSTB affected mice with collagen-induced arthritis (CIA) and RAW264.7 cells induced with lipopolysaccharide (LPS). Results: The findings show that YSTB could significantly improve the clinical arthritic symptoms of CIA mice (mitigate paw swelling, arthritis score, thymus and spleen indices, augment body weight), downregulated expression of pro-inflammatory cytokines like tumor necrosis factor-alpha (TNF-α), interleukin-1ß (IL-1ß), IL-6 and IL-17, while upregulated the level of anti-inflammatory like IL-10 and transforming growth factor-ß (TGF-ß). Meanwhile, YSTB inhibits bone erosion and reduces inflammatory cell infiltration, synovial proliferation, and joint destruction in CIA mice. In addition, we found that YSTB was able to suppress the LPS-induced inflammation of RAW264.7 cells, which was ascribed to the suppression of nitric oxide (NO) production and reactive oxygen species formation (ROS). YSTB also inhibited the production of inducible nitric oxide synthase and reduced the releases of pro-inflammatory cytokines TNF-α, IL-1ß, and IL-6 in LPS-induced RAW264.7 cells. Furthermore, the phosphorylation expression of JAK2, JAK3, STAT3, p38, ERK and p65 protein could be suppressed by YSTB, while the expression of SOCS3 could be activated. Conclusion: Taken together, YSTB possesses anti-inflammatory and prevention bone destruction effects in RA disease by regulating the JAK/STAT3/SOCS3 signaling pathway.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Drugs, Chinese Herbal , Janus Kinases , STAT3 Transcription Factor , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein , Animals , Mice , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , RAW 264.7 Cells , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Arthritis, Experimental/metabolism , Signal Transduction/drug effects , Janus Kinases/metabolism , Male , Cytokines/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Mice, Inbred DBA , Disease Models, Animal
3.
Biochemistry (Mosc) ; 89(6): 1024-1030, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38981698

ABSTRACT

Tyrosine hydroxylase (TH) catalyzes hydroxylation of L-tyrosine to L-3,4-dihydroxyphenylalanine, the initial and rate-limiting step in the synthesis of dopamine, noradrenaline, and adrenaline. Mutations in the human TH gene are associated with hereditary motor disorders. The common C886T mutation identified in the mouse Th gene results in the R278H substitution in the enzyme molecule. We investigated the impact of this mutation on the TH activity in the mouse midbrain. The TH activity in the midbrain of Mus musculus castaneus (CAST) mice homozygous for the 886C allele was higher compared to C57BL/6 and DBA/2 mice homozygous for the 886T allele. Notably, this difference in the enzyme activity was not associated with changes in the Th gene mRNA levels and TH protein content. Analysis of the TH activity in the midbrain in mice from the F2 population obtained by crossbreeding of C57BL/6 and CAST mice revealed that the 886C allele is associated with a high TH activity. Moreover, this allele showed complete dominance over the 886T allele. However, the C886T mutation did not affect the levels of TH protein in the midbrain. These findings demonstrate that the C886T mutation is a major genetic factor determining the activity of TH in the midbrain of common laboratory mouse strains. Moreover, it represents the first common spontaneous mutation in the mouse Th gene whose influence on the enzyme activity has been demonstrated. These results will help to understand the role of TH in the development of adaptive and pathological behavior, elucidate molecular mechanisms regulating the activity of TH, and explore pharmacological agents for modulating its function.


Subject(s)
Mice, Inbred C57BL , Tyrosine 3-Monooxygenase , Animals , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Mice , Mutation , Brain/metabolism , Mice, Inbred DBA , Mesencephalon/metabolism , Mesencephalon/enzymology , Male , Alleles
4.
J Nippon Med Sch ; 91(3): 285-295, 2024.
Article in English | MEDLINE | ID: mdl-38972741

ABSTRACT

BACKGROUND: The standard treatment for Kawasaki disease is immunoglobulin therapy, but the high frequency of coronary sequelae in immunoglobulin-refractory cases indicates a need for further improvement in treatment. METHODS: Kawasaki disease-like vasculitis was induced in 5-week-old DBA/2 mice by intraperitoneal administration of 0.5 mg Candida albicans water-soluble fraction (CAWS) daily for 5 days followed by daily administration of candesartan, an angiotensin receptor blocker. The vasculitis suppression effect was confirmed histologically and serologically in mice sacrificed at 28 days after the start of candesartan. RESULTS: The area of inflammatory cell infiltration at the aortic root was 2.4±1.4% in the Control group, 18.1±1.9% in the CAWS group, and 7.1±2.3%, 5.8±1.4%, 7.6±2.4%, and 7.9±5.0% in the CAWS+candesartan 0.125-mg/kg, 0.25-mg/kg, 0.5-mg/kg, and 1.0-mg/kg groups, respectively (p=0.0200, p=0.0122, p=0.0122, and p=0.0200 vs. CAWS, respectively). The low-dose candesartan group also showed significantly reduced inflammatory cell infiltration. A similar trend was confirmed by immunostaining of macrophages and TGFß receptors. Measurement of the inflammatory cytokines IL-1ß, IL-6, and TNF-α confirmed the anti-vasculitis effect of candesartan. CONCLUSIONS: Candesartan inhibited vasculitis even at clinical doses used in children, making it a strong future candidate as an additional treatment for immunoglobulin-refractory Kawasaki disease.


Subject(s)
Benzimidazoles , Biphenyl Compounds , Candida albicans , Disease Models, Animal , Mucocutaneous Lymph Node Syndrome , Tetrazoles , Animals , Benzimidazoles/pharmacology , Benzimidazoles/administration & dosage , Mucocutaneous Lymph Node Syndrome/drug therapy , Tetrazoles/pharmacology , Tetrazoles/administration & dosage , Candida albicans/drug effects , Biphenyl Compounds/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Mice, Inbred DBA , Solubility , Water , Vasculitis/drug therapy , Male , Mice , Cytokines/metabolism , Interleukin-6/metabolism
5.
Commun Biol ; 7(1): 804, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961129

ABSTRACT

Oral administration of harmless antigens can induce suppression of reactive immune responses, a process that capitalises on the ability of the gastrointestinal tract to tolerate exposure to food and commensal microbiome without triggering inflammatory responses. Repeating exposure to type II collagen induces oral tolerance and inhibits induction of arthritis, a chronic inflammatory joint condition. Although some mechanisms underlying oral tolerance are described, how dysregulation of gut immune networks impacts on inflammation of distant tissues like the joints is unclear. We used undenatured type II collagen in a prophylactic regime -7.33 mg/kg three times/week- to describe the mechanisms associated with protective oral immune-therapy (OIT) in gut and joint during experimental Collagen-Induced Arthritis (CIA). OIT reduced disease incidence to 50%, with reduced expression of IL-17 and IL-22 in the joints of asymptomatic mice. Moreover, whilst the gut tissue of arthritic mice shows substantial damage and activation of tissue-specific immune networks, oral administration of undenatured type II collagen protects against gut pathology in all mice, symptomatic and asymptomatic, rewiring IL-17/IL-22 networks. Furthermore, gut fucosylation and microbiome composition were also modulated. These results corroborate the relevance of the gut-joint axis in arthritis, showing novel regulatory mechanisms linked to therapeutic OIT in joint disease.


Subject(s)
Arthritis, Experimental , Collagen Type II , Gastrointestinal Microbiome , Homeostasis , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/prevention & control , Collagen Type II/immunology , Mice , Gastrointestinal Microbiome/drug effects , Male , Joints/immunology , Joints/drug effects , Joints/pathology , Mice, Inbred DBA , Interleukin-17/metabolism , Interleukin-22 , Administration, Oral
6.
Int J Mol Sci ; 25(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38892148

ABSTRACT

The primary emphasis of photoimmunology is the impact of nonionizing radiation on the immune system. With the development of terahertz (THz) and sub-terahertz (sub-THz) technology, the biological effects of this emerging nonionizing radiation, particularly its influence on immune function, remain insufficiently explored but are progressively attracting attention. Here, we demonstrated that 0.1 sub-THz radiation can modulate the immune system and alleviate symptoms of arthritis in collagen-induced arthritis (CIA) mice through a nonthermal manner. The application of 0.1 sub-THz irradiation led to a decrease in proinflammatory factors within the joints and serum, reducing the levels of blood immune cells and the quantity of splenic CD4+ T cells. Notably, 0.1 sub-THz irradiation restored depleted Treg cells in CIA mice and re-established the Th17/Treg equilibrium. These findings suggested that sub-THz irradiation plays a crucial role in systemic immunoregulation. Further exploration of its immune modulation mechanisms revealed the anti-inflammatory properties of 0.1 sub-THz on LPS-stimulated skin keratinocytes. Through the reduction in NF-κB signaling and NLRP3 inflammasome activation, 0.1 sub-THz irradiation effectively decreased the production of inflammatory factors and immune-active substances, including IL-1ß and PGE2, in HaCaT cells. Consequently, 0.1 sub-THz irradiation mitigated the inflammatory response and contributed to the maintenance of immune tolerance in CIA mice. This research provided significant new evidence supporting the systemic impacts of 0.1 sub-THz radiation, particularly on the immune system. It also enhanced the field of photoimmunology and offered valuable insights into the potential biomedical applications of 0.1 sub-THz radiation for treating autoimmune diseases.


Subject(s)
Arthritis, Experimental , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/radiotherapy , Arthritis, Experimental/pathology , Mice , Terahertz Radiation , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Inflammasomes/immunology , NF-kappa B/metabolism , Mice, Inbred DBA , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/radiation effects , Humans , Signal Transduction/radiation effects , Keratinocytes/radiation effects , Keratinocytes/immunology , Keratinocytes/metabolism
7.
Int J Biol Sci ; 20(8): 2922-2942, 2024.
Article in English | MEDLINE | ID: mdl-38904021

ABSTRACT

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovial inflammation and the production of autoantibodies. Previous studies have indicated an association between high-salt diets (HSD) and an increased risk of RA, yet the underlying mechanisms remain unclear. Macrophage pyroptosis, a pro-inflammatory form of cell death, plays a pivotal role in RA. In this study, we demonstrate that HSD exacerbates the severity of arthritis in collagen-induced arthritis (CIA) mice, correlating with macrophage infiltration and inflammatory lesions. Given the significant alterations observed in macrophages from CIA mice subjected to HSD, we specifically investigate the impact of HSD on macrophage responses in the inflammatory milieu of RA. In our in vitro experiments, pretreatment with NaCl enhances LPS-induced pyroptosis in RAW.264.7 and THP-1 cells through the p38 MAPK/NF-κB signaling pathway. Subsequent experiments reveal that Slc6a12 inhibitors and SGK1 silencing inhibit sodium-induced activation of macrophage pyroptosis and the p38 MAPK/NF-κB signaling pathway, whereas overexpression of the SGK1 gene counteracts the effect of sodium on macrophages. In conclusion, our findings verified that high salt intake promotes the progression of RA and provided a detailed elucidation of the activation of macrophage pyroptosis induced by sodium transportation through the Slc6a12 channel.


Subject(s)
Arthritis, Rheumatoid , Macrophages , Protein Serine-Threonine Kinases , Pyroptosis , Animals , Mice , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Macrophages/metabolism , Pyroptosis/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Sodium Chloride/pharmacology , RAW 264.7 Cells , Humans , Male , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/genetics , Arthritis, Experimental/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Mice, Inbred DBA
8.
Clin Exp Hypertens ; 46(1): 2366270, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38864268

ABSTRACT

OBJECTIVE: To elucidate the underlying mechanism by which the proliferation and migration abilities of human umbilical cord mesenchymal stem cells (hUC-MSCs) determine their therapeutic efficacy in rheumatoid arthritis treatment. METHODS: The DBA/1J mice were utilized to establish a collagen-induced RA (CIA) mouse model and to validate the therapeutic efficacy of hUC-MSCs transfected with CD151 siRNA. RNA-seq, QT-PCR and western blotting were utilized to evaluate the mRNA and protein levels of the PI3K/AKT pathway, respectively. RESULTS: IFN-γ significantly enhanced the proliferation and migration abilities of hUC-MSCs, up-regulating the expression of CD151, a gene related to cell proliferation and migration. Effective inhibition of this effect was achieved through CD151 siRNA treatment. However, IFN-γ did not affect hUC-MSCs differentiation or changes in cell surface markers. Additionally, transplantation of CD151-interfered hUC-MSCs (siRNA-CD151-hUC-MSCs) resulted in decreased colonization in the toes of CIA mice and worse therapeutic effects compared to empty vector treatment (siRNA-NC-hUC-MSCs). CONCLUSION: IFN-γ facilitates the proliferation and migration of hUC-MSCs through the CD151/PI3K/AKT pathway. The therapeutic efficacy of siRNA-CD151-hUC-MSCs was found to be inferior to that of siRNA-NC-hUC-MSCs.


Subject(s)
Arthritis, Rheumatoid , Cell Movement , Cell Proliferation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Mice, Inbred DBA , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Arthritis, Rheumatoid/therapy , Arthritis, Rheumatoid/metabolism , Mice , Mesenchymal Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Mesenchymal Stem Cell Transplantation/methods , Phosphatidylinositol 3-Kinases/metabolism , Humans , Interferon-gamma/metabolism , Umbilical Cord/cytology , Arthritis, Experimental/therapy , Arthritis, Experimental/metabolism , Male
9.
Int Immunopharmacol ; 136: 112383, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38843642

ABSTRACT

The treatment of autoimmune and inflammatory diseases often requires targeting multiple pathogenic pathways. KYS202004A is a novel bispecific fusion protein designed to antagonize TNF-α and IL-17A, pivotal in the pathophysiology of autoimmune and inflammatory diseases. Our initial efforts focused on screening for optimal structure by analyzing expression levels, purity, and binding capabilities. The binding affinity of KYS202004A to TNF-α and IL-17A was evaluated using SPR. In vitro, we assessed the inhibitory capacity of KYS202004A on cytokine-induced CXCL1 expression in HT29 cells. In vivo, its efficacy was tested using a Collagen-Induced Arthritis (CIA) model in transgenic human-IL-17A mice and an imiquimod-induced psoriasis model in cynomolgus monkeys. KYS202004A demonstrated significant inhibition of IL-17A and TNF-α signaling pathways, outperforming the efficacy of monotherapeutic agents ixekizumab and etanercept in reducing CXCL1 expression in vitro and ameliorating disease markers in vivo. In the CIA model, KYS202004A significantly reduced clinical symptoms, joint destruction, and serum IL-6 concentrations. The psoriasis model revealed that KYS202004A, particularly at a 2  mg/kg dose, was as effective as the combination of ixekizumab and etanercept. This discovery represents a significant advancement in treating autoimmune and inflammatory diseases, offering a dual-targeted therapeutic approach with enhanced efficacy over current monotherapies.


Subject(s)
Arthritis, Experimental , Interleukin-17 , Macaca fascicularis , Psoriasis , Recombinant Fusion Proteins , Tumor Necrosis Factor-alpha , Animals , Interleukin-17/metabolism , Tumor Necrosis Factor-alpha/metabolism , Humans , Psoriasis/drug therapy , Psoriasis/immunology , Psoriasis/chemically induced , Recombinant Fusion Proteins/therapeutic use , Recombinant Fusion Proteins/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Mice , Chemokine CXCL1/metabolism , Chemokine CXCL1/genetics , HT29 Cells , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Mice, Transgenic , Disease Models, Animal , Antibodies, Bispecific/therapeutic use , Antibodies, Bispecific/pharmacology , Male , Drug Evaluation, Preclinical , Imiquimod , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Mice, Inbred DBA
10.
J Ethnopharmacol ; 333: 118422, 2024 Oct 28.
Article in English | MEDLINE | ID: mdl-38838922

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Traditional Chinese herbal medicine Panax japonicus C.A. Meyer has a long history in clinical treatment of rheumatoid arthritis (RA). Total saponins of Panax japonicus C.A. Meyer (TSPJs) were extracted from the root of Panax japonicus C.A. Meyer, and its anti-rheumatism mechanism is still unclear. AIM OF THE STUDY: To investigate whether TSPJs attenuated synovial angiogenesis in RA and explore the potential mechanisms. MATERIALS AND METHODS: Potential TSPJs targets involving gene function were predicted by network pharmacology related databases. Bioinformatics analysis and molecular docking technology were used to predict the mechanism of TSPJs in the treatment of RA. The predicted results were validated by cell experiments and a collagen-induced arthritis (CIA) mouse model. RESULTS: Bioinformatics analysis results showed that TSPJs may inhibit RA-related angiogenesis through the hypoxia-inducible factor-1 (HIF-1) and vascular endothelial growth factor (VEGF) pathways. In vitro, different doses of TSPJs showed a good inhibitory effect on the tube formation of EA.hy926 cells. The results of the cellular thermal shift assay indicated that TSPJs can bind to the HIF-1α, VEGFA, and angiopoietin-1 (ANG-1) proteins. In vivo, the administration of TSPJs alleviated the symptoms of CIA mice, including the arthritis index, hind paw thickness, and swollen joint count. The histological results demonstrated that TSPJs inhibited inflammation, angiogenesis, bone damage, and cartilage destruction. Furthermore, TSPJs decreased the number of vessels and the expression level of CD31. The mechanistic results revealed that TSPJs decreased the expression of HIF-1α, VEGFA, and ANG-1 in the serum or synovial tissues of CIA mice. CONCLUSION: These results suggest that TSPJs effectively inhibit angiogenesis in RA, and the mechanism may be related to inhibiting the HIF-1α/VEGF/ANG-1 axis.


Subject(s)
Angiogenesis Inhibitors , Angiopoietin-1 , Arthritis, Experimental , Arthritis, Rheumatoid , Hypoxia-Inducible Factor 1, alpha Subunit , Panax , Saponins , Vascular Endothelial Growth Factor A , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Saponins/pharmacology , Saponins/therapeutic use , Arthritis, Rheumatoid/drug therapy , Vascular Endothelial Growth Factor A/metabolism , Panax/chemistry , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Humans , Angiogenesis Inhibitors/pharmacology , Male , Mice , Angiopoietin-1/metabolism , Molecular Docking Simulation , Mice, Inbred DBA , Neovascularization, Pathologic/drug therapy , Signal Transduction/drug effects , Plant Roots/chemistry
11.
eNeuro ; 11(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38937109

ABSTRACT

Elevated intraocular pressure (IOP) triggers glaucoma by damaging the output neurons of the retina called retinal ganglion cells (RGCs). This leads to the loss of RGC signaling to visual centers of the brain such as the dorsolateral geniculate nucleus (dLGN), which is critical for processing and relaying information to the cortex for conscious vision. In response to altered levels of activity or synaptic input, neurons can homeostatically modulate postsynaptic neurotransmitter receptor numbers, allowing them to scale their synaptic responses to stabilize spike output. While prior work has indicated unaltered glutamate receptor properties in the glaucomatous dLGN, it is unknown whether glaucoma impacts dLGN inhibition. Here, using DBA/2J mice, which develop elevated IOP beginning at 6-7 months of age, we tested whether the strength of inhibitory synapses on dLGN thalamocortical relay neurons is altered in response to the disease state. We found an enhancement of feedforward disynaptic inhibition arising from local interneurons along with increased amplitude of quantal inhibitory synaptic currents. A combination of immunofluorescence staining for the γ-aminobutyric acid (GABA)A-α1 receptor subunit, peak-scaled nonstationary fluctuation analysis, and measures of homeostatic synaptic scaling pointed to an ∼1.4-fold increase in GABA receptors at postsynaptic inhibitory synapses, although several pieces of evidence indicate a nonuniform scaling across inhibitory synapses within individual relay neurons. Together, these results indicate an increase in inhibitory synaptic strength in the glaucomatous dLGN, potentially pointing toward homeostatic compensation for disruptions in network and neuronal function triggered by increased IOP.


Subject(s)
Disease Models, Animal , Geniculate Bodies , Glaucoma , Mice, Inbred DBA , Neural Inhibition , Synapses , Animals , Geniculate Bodies/physiology , Glaucoma/metabolism , Glaucoma/physiopathology , Glaucoma/pathology , Neural Inhibition/physiology , Synapses/physiology , Synapses/metabolism , Male , Inhibitory Postsynaptic Potentials/physiology , Mice , Female , Intraocular Pressure/physiology , Receptors, GABA-A/metabolism
12.
Int J Mol Sci ; 25(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38892051

ABSTRACT

Dietary supplementation with n-3 polyunsaturated fatty acids (PUFA) has been found to be beneficial in rodent rheumatoid arthritis models and human trials. However, the molecular targets of n-3 PUFAs and their beneficial effects on rheumatoid arthritis are under-researched. Free fatty acid receptor 4 (FFA4, also known as GPR120) is a receptor for n-3 PUFA. We aim to investigate whether FFA4 activation reduces collagen-induced rheumatoid arthritis (CIA) by using an FFA4 agonist, compound A (CpdA), in combination with DBA-1J Ffa4 gene wild-type (WT) and Ffa4 gene knock-out (KO) mice. CIA induced an increase in the arthritis score, foot edema, synovial hyperplasia, pannus formation, proteoglycan loss, cartilage damage, and bone erosion, whereas the administration of CpdA significantly suppressed those increases in Ffa4 WT mice but not Ffa4 gene KO mice. CIA increased mRNA expression levels of pro-inflammatory Th1/Th17 cytokines, whereas CpdA significantly suppressed those increases in Ffa4 WT mice but not Ffa4 gene KO mice. CIA induced an imbalance between Th1/Th17 and Treg cells, whereas CpdA rebalanced them in spleens from Ffa4 WT mice but not Ffa4 gene KO mice. In SW982 synovial cells, CpdA reduced the LPS-induced increase in pro-inflammatory cytokine levels. In summary, the present results suggest that the activation of FFA4 in immune and synovial cells could suppress the characteristics of rheumatoid arthritis and be an adjuvant therapy.


Subject(s)
Arthritis, Experimental , Mice, Knockout , Receptors, G-Protein-Coupled , T-Lymphocytes, Regulatory , Th1 Cells , Th17 Cells , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Arthritis, Experimental/drug therapy , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/agonists , Mice , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/drug effects , Mice, Inbred DBA , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Male , Cytokines/metabolism
13.
Arthritis Res Ther ; 26(1): 121, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879555

ABSTRACT

BACKGROUND: Janus kinase (JAK) inhibitors, such as baricitinib, are widely used to treat rheumatoid arthritis (RA). Clinical studies show that baricitinib is more effective at reducing pain than other similar drugs. Here, we aimed to elucidate the molecular mechanisms underlying the pain relief conferred by baricitinib, using a mouse model of arthritis. METHODS: We treated collagen antibody-induced arthritis (CAIA) model mice with baricitinib, celecoxib, or vehicle, and evaluated the severity of arthritis, histological findings of the spinal cord, and pain-related behaviours. We also conducted RNA sequencing (RNA-seq) to identify alterations in gene expression in the dorsal root ganglion (DRG) following baricitinib treatment. Finally, we conducted in vitro experiments to investigate the direct effects of baricitinib on neuronal cells. RESULTS: Both baricitinib and celecoxib significantly decreased CAIA and improved arthritis-dependent grip-strength deficit, while only baricitinib notably suppressed residual tactile allodynia as determined by the von Frey test. CAIA induction of inflammatory cytokines in ankle synovium, including interleukin (IL)-1ß and IL-6, was suppressed by treatment with either baricitinib or celecoxib. In contrast, RNA-seq analysis of the DRG revealed that baricitinib, but not celecoxib, restored gene expression alterations induced by CAIA to the control condition. Among many pathways changed by CAIA and baricitinib treatment, the interferon-alpha/gamma, JAK-signal transducer and activator of transcription 3 (STAT3), and nuclear factor kappa B (NF-κB) pathways were considerably decreased in the baricitinib group compared with the celecoxib group. Notably, only baricitinib decreased the expression of colony-stimulating factor 1 (CSF-1), a potent cytokine that causes neuropathic pain through activation of the microglia-astrocyte axis in the spinal cord. Accordingly, baricitinib prevented increases in microglia and astrocytes caused by CAIA. Baricitinib also suppressed JAK/STAT3 pathway activity and Csf1 expression in cultured neuronal cells. CONCLUSIONS: Our findings demonstrate the effects baricitinib has on the DRG in relation to ameliorating both inflammatory and neuropathic pain.


Subject(s)
Arthritis, Experimental , Ganglia, Spinal , Interleukin-6 , Neuralgia , STAT3 Transcription Factor , Signal Transduction , Animals , Male , Mice , Arthritis, Experimental/metabolism , Arthritis, Experimental/drug therapy , Azetidines/pharmacology , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Interleukin-6/metabolism , Janus Kinase Inhibitors/pharmacology , Mice, Inbred DBA , Neuralgia/drug therapy , Neuralgia/metabolism , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , Purines/pharmacology , Pyrazoles/pharmacology , Signal Transduction/drug effects , STAT3 Transcription Factor/metabolism , Sulfonamides/pharmacology
14.
Int J Mol Sci ; 25(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38928413

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease that significantly impacts quality of life by disrupting CD4+ T cell immune homeostasis. The identification of a low-side-effect drug for RA treatment is urgently needed. Our previous study suggests that Trichinella spiralis paramyosin (Ts-Pmy) has immunomodulatory effects, but its potential effect on CD4+ T cell response in RA remains unclear. In this study, we used a murine model to investigate the role of rTs-Pmy in regulating CD4+ T cell differentiation in collagen-induced arthritis (CIA). Additionally, we assessed the impact of rTs-Pmy on CD4+ T cell differentiation towards the Th1 and Th17 phenotypes, which are associated with inflammatory responses in arthritis, using in vitro assays. The results demonstrated that rTs-Pmy administration reduced arthritis severity by inhibiting Th1 and Th17 response while enhancing Treg response. Prophylactic administration of Ts-Pmy showed superior efficacy on CIA compared to therapeutic administration. Furthermore, in vitro assays demonstrated that rTs-Pmy could inhibit the differentiation of CD4+ T cells into Th1 and Th17 while inducing the production of Tregs, suggesting a potential mechanism underlying its therapeutic effects. This study suggests that Ts-Pmy may ameliorate CIA by restoring the immune balance of CD4+ T cells and provides new insights into the mechanism through which helminth-derived proteins exert their effects on autoimmune diseases.


Subject(s)
Arthritis, Experimental , CD4-Positive T-Lymphocytes , Cell Differentiation , Th17 Cells , Trichinella spiralis , Tropomyosin , Animals , Trichinella spiralis/immunology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Experimental/drug therapy , Mice , Cell Differentiation/drug effects , Tropomyosin/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th1 Cells/immunology , Male , Helminth Proteins/pharmacology , Helminth Proteins/therapeutic use , Helminth Proteins/immunology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/drug therapy , T-Lymphocytes, Regulatory/immunology , Disease Models, Animal , Mice, Inbred DBA
15.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 73-77, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38836679

ABSTRACT

GABBR1 receptors have been implicated in the progression of rheumatoid arthritis (RA), and p38 MAP kinase (MAPK) was shown to be downregulated by GABA and result in unchecked production of pro-inflammatory cytokine. GABBR1 is a member of GABA receptors, and it is known to be upregulated and plays a vital role in RA. Glucocorticoids are efficient therapeutics in rheumatoid arthritis (RA) and are known to regulate GABA actions; therefore, we intended to investigate the potential of glucocorticoids in RA concerning the potential pathway GABBR1/MAPK. Joint specimens were obtained from collagen-induced arthritis mouse model. A double-blind semi-quantitative analysis of vascularity, cell infiltration, as well as lining thickness by help of a 4-point scale setting was used to assess joint inflammation. Expression of GABBR1 and p38 was evaluated immunohistochemically. In vitro peripheral blood (PB), synovial fluid (SF), and mononuclear cells (MCs) were acquired from RA mice. Western blotting was used for detecting expression of GABBR1 and p38 proteins. The presence of high levels of GABBR1 and p38 was prevalent in RA joints relative to healthy joints and related to the inflammation level. Glucocorticoid treatment alters GABBR1 along with p38 protein expression in joints while reducing joint inflammation. Ex vivo and in vitro assays revealed glucocorticoids have a direct impact on p38, such as the decreased GABBR1 expression level after dexamethasone incubation with SFMC. GABBR1 together with p38 expression in RA joints depends on local inflammation and can be targeted by glucocorticoids.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Glucocorticoids , p38 Mitogen-Activated Protein Kinases , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Animals , Glucocorticoids/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Mice , Male , Joints/pathology , Joints/drug effects , Joints/metabolism , Mice, Inbred DBA , Synovial Fluid/metabolism , Synovial Fluid/drug effects , Cellular Microenvironment/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/drug effects , Disease Models, Animal
16.
BMC Musculoskelet Disord ; 25(1): 447, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844896

ABSTRACT

BACKGROUND: Although various anti-inflammatory medicines are widely recommended for osteoarthritis (OA) treatment, no significantly clinical effect has been observed. This study aims to examine the effects of vitamin B6, a component that has been reported to be capable of alleviating inflammation and cell death in various diseases, on cartilage degeneration in OA. METHODS: Collagen-induced arthritis (CIA) mice model were established and the severity of OA in cartilage was determined using the Osteoarthritis Research Society International (OARSI) scoring system. The mRNA and protein levels of indicators associated with extracellular matrix (ECM) metabolism, apoptosis and inflammation were detected. The effect of vitamin B6 (VB6) on the mice were assessed using HE staining and masson staining. The apoptosis rate of cells was assessed using TdT-mediated dUTP nick end labeling. RESULTS: Our results showed a trend of improved OARSI score in mice treated with VB6, which remarkably inhibited the hyaline cartilage thickness, chondrocyte disordering, and knees hypertrophy. Moreover, the VB6 supplementation reduced the protein expression of pro-apoptosis indicators, including Bax and cleaved caspase-3 and raised the expression level of anti-apoptosis marker Bcl-2. Importantly, VB6 improved ECM metabolism in both in vivo and in vitro experiments. CONCLUSIONS: This study demonstrated that VB6 alleviates OA through regulating ECM metabolism, inflammation and apoptosis in chondrocytes and CIA mice. The findings in this study provide a theoretical basis for targeted therapy of OA, and further lay the theoretical foundation for studies of mechanisms of VB6 in treating OA.


Subject(s)
Apoptosis , Arthritis, Experimental , Chondrocytes , Inflammation , Osteoarthritis , Vitamin B 6 , Animals , Apoptosis/drug effects , Mice , Vitamin B 6/pharmacology , Vitamin B 6/therapeutic use , Osteoarthritis/drug therapy , Osteoarthritis/pathology , Osteoarthritis/metabolism , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Arthritis, Experimental/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Male , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Mice, Inbred DBA , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/pathology , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cartilage, Articular/metabolism
17.
J Ethnopharmacol ; 332: 118286, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-38723919

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Di-Long (Pheretima vulgaris) is a classic animal sourced traditional Chinese medicine. It has been used for the treatment of joint inflammation and arthralgia for over two thousand years due to its effects of Tong-Luo-Zhi-Tong (dredging collaterals and alleviating pain). Our previous study showed that Chinese medicine Di-Long has significant anti-rheumatoid arthritis (RA) effects. AIM OF THE STUDY: Considering Di-Long as a potential source of active compounds with specific anti-RA therapeutic effects, this research was to obtain the anti-RA target-specific active fraction from Di-Long extracts (DL), and to further explore the chemical basis and verify the anti-RA mechanism of this active fraction. MATERIALS AND METHODS: Transcriptomic was applied to obtain the main anti-RA targets of DL on human RA fibroblast-like synoviocytes (FLS) and validated by qPCR. The target-corresponding active fraction was isolated from DL by ethanol precipitation and gel chromatography, and analyzed by nanoliter chromatography-mass spectrometry. Anti-RA effects of this active fraction was investigated by collagen-induced arthritis (CIA) in mice, and anti-RA mechanisms were verified in cocultured model of rat FLS and peripheral blood lymphocytes. RESULTS: We confirmed that CXCL10/CXCR3 was the main anti-RA target of DL. The active fraction - A (2182 - 890 Da) was isolated from DL based on its CXCL10 inhibiting effects in RA-FLS. Fraction A contains 195 peptides (192 were newly discovered), 26 of which might be bioactive and were considered to be the chemical basis of its anti-RA effects. Fraction A significantly ameliorated the joint destruction and overall inflammation in CIA mice, and downregulated CXCR3 expression in mice joint. Fraction A inhibited the chemotaxis of Th-cells in rat peripheral blood lymphocytes towards the TNF-α-induced rat FLS through CXCL10/CXCR3 pathway. CONCLUSIONS: Our work indicated that active fraction from DL containing small peptides exhibits promising therapeutic effects for RA through inhibiting CXCL10/CXCR3 chemotaxis.


Subject(s)
Antirheumatic Agents , Arthritis, Experimental , Arthritis, Rheumatoid , Chemokine CXCL10 , Chemotaxis , Receptors, CXCR3 , Synovial Membrane , Animals , Receptors, CXCR3/metabolism , Chemokine CXCL10/metabolism , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Male , Antirheumatic Agents/pharmacology , Antirheumatic Agents/isolation & purification , Rats , Humans , Chemotaxis/drug effects , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Mice , Mice, Inbred DBA , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Synoviocytes/drug effects , Synoviocytes/metabolism
18.
Clin Immunol ; 264: 110255, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763433

ABSTRACT

Fibroblast-like synoviocytes (FLS) play critical roles in rheumatoid arthritis (RA). Itaconate (ITA), an endogenous metabolite derived from the tricarboxylic acid (TCA) cycle, has attracted attention because of its anti-inflammatory, antiviral, and antimicrobial effects. This study evaluated the effect of ITA on FLS and its potential to treat RA. ITA significantly decreased FLS proliferation and migration in vitro, as well as mitochondrial oxidative phosphorylation and glycolysis measured by an extracellular flux analyzer. ITA accumulates metabolites including succinate and citrate in the TCA cycle. In rats with type II collagen-induced arthritis (CIA), intra-articular injection of ITA reduced arthritis and bone erosion. Irg1-deficient mice lacking the ability to produce ITA had more severe arthritis than control mice in the collagen antibody-induced arthritis. ITA ameliorated CIA by inhibiting FLS proliferation and migration. Thus, ITA may be a novel therapeutic agent for RA.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Cell Movement , Cell Proliferation , Fibroblasts , Succinates , Synoviocytes , Animals , Synoviocytes/drug effects , Synoviocytes/metabolism , Cell Movement/drug effects , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Cell Proliferation/drug effects , Succinates/pharmacology , Rats , Fibroblasts/drug effects , Fibroblasts/metabolism , Male , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Mice , Mice, Knockout , Cells, Cultured , Mice, Inbred DBA , Citric Acid Cycle/drug effects
19.
Int Immunopharmacol ; 136: 112264, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38810308

ABSTRACT

BACKGROUND: Chemotaxis and trafficking of dendritic cells (DCs) induced by cytokine receptors are crucial steps in rheumatoid arthritis (RA) pathogenesis. C-C chemokine receptor type 5 (CCR5) plays a key role in DC movement and has been implicated in multitudinous inflammatory and immunology diseases. Thus, targeting CCR5 to suppress DC chemotaxis is considered as a potential strategy for the management of RA. METHODS: Herein, we first synthesized a new hybrid named CT3-1 which based on artesunate and isatin. Besides, we studied the regulating effectiveness of CT3-1 on bone marrow-derived DCs (BMDCs) and on collagen-induced arthritis (CIA) through RNA-seq analysis, cell function experiments in vitro and mice model in vivo. RESULTS: The results shown that CT3-1 mainly reduced CCR5 expression of immature BMDCs and importantly inhibited immature BMDC migration induced by CCR5 in vitro, with no or minor influence on other functions of DCs, such as phagocytosis and maturation. In the mouse model, CT3-1 relieved arthritis severity and inhibited CIA development. Furthermore, CT3-1 intervention decreased the expression of CCR5 in DCs and reduced the proportion of DCs in the peripheral blood of CIA mice. CONCLUSIONS: Our findings suggest that CCR5-induced chemotaxis and trafficking of immature DCs are important in RA. Targeting CCR5 and inhibiting immature DC chemotaxis may provide a novel choice for the treatment of RA and other similar autoimmune diseases. Moreover, we synthesized a new hybrid compound CT3-1 that could inhibit immature DC trafficking and effectively relieve RA by directly reducing the CCR5 expression of immature DCs.


Subject(s)
Artesunate , Arthritis, Experimental , Arthritis, Rheumatoid , Chemotaxis , Dendritic Cells , Receptors, CCR5 , Animals , Dendritic Cells/drug effects , Dendritic Cells/immunology , Receptors, CCR5/metabolism , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Chemotaxis/drug effects , Artesunate/pharmacology , Artesunate/therapeutic use , Mice , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Mice, Inbred DBA , Male , Cells, Cultured , Humans
20.
Bone Res ; 12(1): 31, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782893

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease. Early studies hold an opinion that gut microbiota is environmentally acquired and associated with RA susceptibility. However, accumulating evidence demonstrates that genetics also shape the gut microbiota. It is known that some strains of inbred laboratory mice are highly susceptible to collagen-induced arthritis (CIA), while the others are resistant to CIA. Here, we show that transplantation of fecal microbiota of CIA-resistant C57BL/6J mice to CIA-susceptible DBA/1J mice confer CIA resistance in DBA/1J mice. C57BL/6J mice and healthy human individuals have enriched B. fragilis than DBA/1J mice and RA patients. Transplantation of B. fragilis prevents CIA in DBA/1J mice. We identify that B. fragilis mainly produces propionate and C57BL/6J mice and healthy human individuals have higher level of propionate. Fibroblast-like synoviocytes (FLSs) in RA are activated to undergo tumor-like transformation. Propionate disrupts HDAC3-FOXK1 interaction to increase acetylation of FOXK1, resulting in reduced FOXK1 stability, blocked interferon signaling and deactivation of RA-FLSs. We treat CIA mice with propionate and show that propionate attenuates CIA. Moreover, a combination of propionate with anti-TNF etanercept synergistically relieves CIA. These results suggest that B. fragilis or propionate could be an alternative or complementary approach to the current therapies.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Gastrointestinal Microbiome , Histone Deacetylases , Mice, Inbred C57BL , Synoviocytes , Animals , Humans , Male , Mice , Arthritis, Experimental/pathology , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/microbiology , Fibroblasts/metabolism , Fibroblasts/drug effects , Forkhead Transcription Factors/metabolism , Gastrointestinal Microbiome/drug effects , Histone Deacetylases/metabolism , Mice, Inbred DBA , Signal Transduction/drug effects , Synoviocytes/metabolism , Synoviocytes/drug effects , Synoviocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL